1
|
Epremyan KK, Mamaev DV, Zvyagilskaya RA. Alzheimer's Disease: Significant Benefit from the Yeast-Based Models. Int J Mol Sci 2023; 24:9791. [PMID: 37372938 DOI: 10.3390/ijms24129791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Alzheimer's disease (AD) is an age-related, multifaceted neurological disorder associated with accumulation of aggregated proteins (amyloid Aβ and hyperphosphorylated tau), loss of synapses and neurons, and alterations in microglia. AD was recognized by the World Health Organization as a global public health priority. The pursuit of a better understanding of AD forced researchers to pay attention to well-defined single-celled yeasts. Yeasts, despite obvious limitations in application to neuroscience, show high preservation of basic biological processes with all eukaryotic organisms and offer great advantages over other disease models due to the simplicity, high growth rates on low-cost substrates, relatively simple genetic manipulations, the large knowledge base and data collections, and availability of an unprecedented amount of genomic and proteomic toolboxes and high-throughput screening techniques, inaccessible to higher organisms. Research reviewed above clearly indicates that yeast models, together with other, more simple eukaryotic models including animal models, C. elegans and Drosophila, significantly contributed to understanding Aβ and tau biology. These models allowed high throughput screening of factors and drugs that interfere with Aβ oligomerization, aggregation and toxicity, and tau hyperphosphorylation. In the future, yeast models will remain relevant, with a focus on creating novel high throughput systems to facilitate the identification of the earliest AD biomarkers among different cellular networks in order to achieve the main goal-to develop new promising therapeutic strategies to treat or prevent the disease.
Collapse
Affiliation(s)
- Khoren K Epremyan
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33/2, 119071 Moscow, Russia
| | - Dmitry V Mamaev
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33/2, 119071 Moscow, Russia
| | - Renata A Zvyagilskaya
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33/2, 119071 Moscow, Russia
| |
Collapse
|
2
|
Dhakal S, Macreadie I. Finding effective combinations of compounds to prevent Alzheimer's disease. Neural Regen Res 2022; 17:2450-2451. [PMID: 35535895 PMCID: PMC9120672 DOI: 10.4103/1673-5374.335812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Sudip Dhakal
- School of Science, Bundoora Campus, RMIT University, Melbourne, VIC, Australia
| | - Ian Macreadie
- School of Science, Bundoora Campus, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
3
|
Chen X, Muñoz-Arellano AJ, Petranovic D. UBB +1 reduces amyloid-β cytotoxicity by activation of autophagy in yeast. Aging (Albany NY) 2021; 13:23953-23980. [PMID: 34751669 PMCID: PMC8610117 DOI: 10.18632/aging.203681] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/02/2021] [Indexed: 04/20/2023]
Abstract
UBB+1 is a mutated version of ubiquitin B peptide caused by a transcriptional frameshift due to the RNA polymerase II "slippage". The accumulation of UBB+1 has been linked to ubiquitin-proteasome system (UPS) dysfunction and neurodegeneration. Alzheimer's disease (AD) is defined as a progressive neurodegeneration and aggregation of amyloid-β peptides (Aβ) is a prominent neuropathological feature of AD. In our previous study, we found that yeast cells expressing UBB+1 at lower level display an increased resistance to cellular stresses under conditions of chronological aging. In order to examine the molecular mechanisms behind, here we performed genome-wide transcriptional analyses and molecular/cellular biology assays. We found that low UBB+1 expression activated the autophagy pathway, increased vacuolar activity, and promoted transport of autophagic marker ATG8p into vacuole. Furthermore, we introduced low UBB+1 expression to our humanized yeast AD models, that constitutively express Aβ42 and Aβ40 peptide, respectively. The co-expression of UBB+1 with Aβ42 or Aβ40 peptide led to reduced intracellular Aβ levels, ameliorated viability, and increased chronological life span. In an autophagy deficient background strain (atg1Δ), intracellular Aβ levels were not affected by UBB+1 expression. Our findings offer insights for reducing intracellular Aβ toxicity via autophagy-dependent cellular pathways under low level of UBB+1 expression.
Collapse
Affiliation(s)
- Xin Chen
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, Gothenburg, Sweden
| | - Ana Joyce Muñoz-Arellano
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Dina Petranovic
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
4
|
Ralph-Epps T, Onu CJ, Vo L, Schmidtke MW, Le A, Greenberg ML. Studying Lipid-Related Pathophysiology Using the Yeast Model. Front Physiol 2021; 12:768411. [PMID: 34777024 PMCID: PMC8581491 DOI: 10.3389/fphys.2021.768411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/04/2021] [Indexed: 01/01/2023] Open
Abstract
Saccharomyces cerevisiae, commonly known as baker's yeast, is one of the most comprehensively studied model organisms in science. Yeast has been used to study a wide variety of human diseases, and the yeast model system has proved to be an especially amenable tool for the study of lipids and lipid-related pathophysiologies, a topic that has gained considerable attention in recent years. This review focuses on how yeast has contributed to our understanding of the mitochondrial phospholipid cardiolipin (CL) and its role in Barth syndrome (BTHS), a genetic disorder characterized by partial or complete loss of function of the CL remodeling enzyme tafazzin. Defective tafazzin causes perturbation of CL metabolism, resulting in many downstream cellular consequences and clinical pathologies that are discussed herein. The influence of yeast research in the lipid-related pathophysiologies of Alzheimer's and Parkinson's diseases is also summarized.
Collapse
Affiliation(s)
- Tyler Ralph-Epps
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Chisom J. Onu
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Linh Vo
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Michael W. Schmidtke
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Anh Le
- Muskegon Catholic Central High School, Muskegon, MI, United States
| | - Miriam L. Greenberg
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| |
Collapse
|
5
|
Dhakal S, Macreadie I. Protein Homeostasis Networks and the Use of Yeast to Guide Interventions in Alzheimer's Disease. Int J Mol Sci 2020; 21:E8014. [PMID: 33126501 PMCID: PMC7662794 DOI: 10.3390/ijms21218014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/24/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's Disease (AD) is a progressive multifactorial age-related neurodegenerative disorder that causes the majority of deaths due to dementia in the elderly. Although various risk factors have been found to be associated with AD progression, the cause of the disease is still unresolved. The loss of proteostasis is one of the major causes of AD: it is evident by aggregation of misfolded proteins, lipid homeostasis disruption, accumulation of autophagic vesicles, and oxidative damage during the disease progression. Different models have been developed to study AD, one of which is a yeast model. Yeasts are simple unicellular eukaryotic cells that have provided great insights into human cell biology. Various yeast models, including unmodified and genetically modified yeasts, have been established for studying AD and have provided significant amount of information on AD pathology and potential interventions. The conservation of various human biological processes, including signal transduction, energy metabolism, protein homeostasis, stress responses, oxidative phosphorylation, vesicle trafficking, apoptosis, endocytosis, and ageing, renders yeast a fascinating, powerful model for AD. In addition, the easy manipulation of the yeast genome and availability of methods to evaluate yeast cells rapidly in high throughput technological platforms strengthen the rationale of using yeast as a model. This review focuses on the description of the proteostasis network in yeast and its comparison with the human proteostasis network. It further elaborates on the AD-associated proteostasis failure and applications of the yeast proteostasis network to understand AD pathology and its potential to guide interventions against AD.
Collapse
Affiliation(s)
| | - Ian Macreadie
- School of Science, RMIT University, Bundoora, Victoria 3083, Australia;
| |
Collapse
|
6
|
Sachsenhauser V, Deng X, Kim HH, Jankovic M, Bardwell JC. Yeast Tripartite Biosensors Sensitive to Protein Stability and Aggregation Propensity. ACS Chem Biol 2020; 15:1078-1088. [PMID: 32105441 DOI: 10.1021/acschembio.0c00083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In contrast to the myriad approaches available to study protein misfolding and aggregation in vitro, relatively few tools are available for the study of these processes in the cellular context. This is in part due to the complexity of the cellular environment which, for instance, interferes with many spectroscopic approaches. Here, we describe a tripartite fusion approach that can be used to assess in vivo protein stability and solubility in the cytosol of Saccharomyces cerevisiae. Our biosensors contain tripartite fusions in which a protein of interest is inserted into antibiotic resistance markers. These fusions act to directly link the aggregation susceptibility and stability of the inserted protein to antibiotic resistance. We demonstrate a linear relationship between the thermodynamic stabilities of variants of the model folding protein immunity protein 7 (Im7) fused into the resistance markers and their antibiotic resistance readouts. We also use this system to investigate the in vivo properties of the yeast prion proteins Sup35 and Rnq1 and proteins whose aggregation is associated with some of the most prevalent neurodegenerative misfolding disorders, including peptide amyloid beta 1-42 (Aβ42), which is involved in Alzheimer's disease, and protein α-synuclein, which is linked to Parkinson's disease.
Collapse
Affiliation(s)
- Veronika Sachsenhauser
- Department of Molecular, Cellular, and Developmental Biology and Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan 48109-1085, United States
- Department of Chemistry, Technical University Munich, Lichtenbergstraße 4, 85748 Garching, Germany
| | - Xiexiong Deng
- Department of Molecular, Cellular, and Developmental Biology and Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan 48109-1085, United States
| | - Hyun-hee Kim
- Department of Molecular, Cellular, and Developmental Biology and Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan 48109-1085, United States
| | - Maja Jankovic
- Department of Molecular, Cellular, and Developmental Biology and Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan 48109-1085, United States
| | - James C.A. Bardwell
- Department of Molecular, Cellular, and Developmental Biology and Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan 48109-1085, United States
| |
Collapse
|
7
|
Chen X, Ji B, Hao X, Li X, Eisele F, Nyström T, Petranovic D. FMN reduces Amyloid-β toxicity in yeast by regulating redox status and cellular metabolism. Nat Commun 2020; 11:867. [PMID: 32054832 PMCID: PMC7018843 DOI: 10.1038/s41467-020-14525-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 01/07/2020] [Indexed: 01/09/2023] Open
Abstract
Alzheimer's disease (AD) is defined by progressive neurodegeneration, with oligomerization and aggregation of amyloid-β peptides (Aβ) playing a pivotal role in its pathogenesis. In recent years, the yeast Saccharomyces cerevisiae has been successfully used to clarify the roles of different human proteins involved in neurodegeneration. Here, we report a genome-wide synthetic genetic interaction array to identify toxicity modifiers of Aβ42, using yeast as the model organism. We find that FMN1, the gene encoding riboflavin kinase, and its metabolic product flavin mononucleotide (FMN) reduce Aβ42 toxicity. Classic experimental analyses combined with RNAseq show the effects of FMN supplementation to include reducing misfolded protein load, altering cellular metabolism, increasing NADH/(NADH + NAD+) and NADPH/(NADPH + NADP+) ratios and increasing resistance to oxidative stress. Additionally, FMN supplementation modifies Htt103QP toxicity and α-synuclein toxicity in the humanized yeast. Our findings offer insights for reducing cytotoxicity of Aβ42, and potentially other misfolded proteins, via FMN-dependent cellular pathways.
Collapse
Affiliation(s)
- Xin Chen
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, SE41296, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, SE41296, Gothenburg, Sweden
| | - Boyang Ji
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, SE41296, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, SE41296, Gothenburg, Sweden
| | - Xinxin Hao
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, SE40530, Gothenburg, Sweden
| | - Xiaowei Li
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, SE41296, Gothenburg, Sweden
| | - Frederik Eisele
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, SE40530, Gothenburg, Sweden
| | - Thomas Nyström
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, SE40530, Gothenburg, Sweden
| | - Dina Petranovic
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, SE41296, Gothenburg, Sweden.
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, SE41296, Gothenburg, Sweden.
| |
Collapse
|
8
|
Fang T, Yan H, Li G, Chen W, Liu J, Jiang L. Chromatin remodeling complexes are involvesd in the regulation of ethanol production during static fermentation in budding yeast. Genomics 2019; 112:1674-1679. [PMID: 31618673 DOI: 10.1016/j.ygeno.2019.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 10/02/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022]
Abstract
The budding yeast Saccharomyces cerevisiae remains a central position among biofuel-producing organisms. However, the gene expression regulatory networks behind the ethanol fermentation is still not fully understood. Using a static fermentation model, we have examined the ethanol yields on biomass of deletion mutants for all yeast nonessential genes encoding transcription factors and their related proteins in the yeast genome. A total of 20 (about 10%) transcription factors are identified to be regulators of ethanol production during fermentation. These transcription factors are mainly involved in cell cycling, chromatin remodeling, transcription, stress response, protein synthesis and lipid synthesis. Our data provides a basis for further understanding mechanisms regulating ethanol production in budding yeast.
Collapse
Affiliation(s)
- Tianshu Fang
- Laboratory for Yeast Molecular and Cell Biology, the Research Center of Fermentation Technology, Department of Food Science, School of Agricultural Engineering and Food Sciences, Shandong University of Technology, Zibo 255000, Shandong Province, China
| | - Hongbo Yan
- Laboratory for Yeast Molecular and Cell Biology, the Research Center of Fermentation Technology, Department of Food Science, School of Agricultural Engineering and Food Sciences, Shandong University of Technology, Zibo 255000, Shandong Province, China
| | - Gaozhen Li
- Laboratory for Yeast Molecular and Cell Biology, the Research Center of Fermentation Technology, Department of Food Science, School of Agricultural Engineering and Food Sciences, Shandong University of Technology, Zibo 255000, Shandong Province, China
| | - Weipeng Chen
- Laboratory for Yeast Molecular and Cell Biology, the Research Center of Fermentation Technology, Department of Food Science, School of Agricultural Engineering and Food Sciences, Shandong University of Technology, Zibo 255000, Shandong Province, China
| | - Jian Liu
- Laboratory for Yeast Molecular and Cell Biology, the Research Center of Fermentation Technology, Department of Food Science, School of Agricultural Engineering and Food Sciences, Shandong University of Technology, Zibo 255000, Shandong Province, China
| | - Linghuo Jiang
- Laboratory for Yeast Molecular and Cell Biology, the Research Center of Fermentation Technology, Department of Food Science, School of Agricultural Engineering and Food Sciences, Shandong University of Technology, Zibo 255000, Shandong Province, China.
| |
Collapse
|
9
|
Simvastatin Efficiently Reduces Levels of Alzheimer's Amyloid Beta in Yeast. Int J Mol Sci 2019; 20:ijms20143531. [PMID: 31330953 PMCID: PMC6678968 DOI: 10.3390/ijms20143531] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/12/2019] [Accepted: 07/17/2019] [Indexed: 11/17/2022] Open
Abstract
A large-scale epidemiology study on statins previously showed that simvastatin was unique among statins in reducing the incidence of dementia. Since amyloid beta (Aβ42) is the protein that is most associated with Alzheimer's disease, this study has focused on how simvastatin influences the turnover of native Aβ42 and Aβ42 fused with green fluorescent protein (GFP), in the simplest eukaryotic model organism, Saccharomyces cerevisiae. Previous studies have established that yeast constitutively producing Aβ42 fused to GFP offer a convenient means of analyzing yeast cellular responses to Aβ42. Young cells clear the GFP fusion protein and do not have green fluorescence while the older population of cells retains the fusion protein and exhibits green fluorescence, offering a fast and convenient means of studying factors that affect Aβ42 turnover. In this study the proportion of cells having GFP fused to Aβ after exposure to simvastatin, atorvastatin and lovastatin was analyzed by flow cytometry. Simvastatin effectively reduced levels of the cellular Aβ42 protein in a dose-dependent manner. Simvastatin promoted the greatest reduction as compared to the other two statins. A comparison with fluconazole, which targets that same pathway of ergosterol synthesis, suggests that effects on ergosterol synthesis do not account for the reduced amounts of Aβ42 fused to GFP. The levels of native Aβ42 following treated with simvastatin were also examined using a more laborious approach, quantitative MALDI TOF mass spectrometry. Simvastatin efficiently reduced levels of native Aβ42 from the population. This work indicates a novel action of simvastatin in reducing levels of Aβ42 providing new insights into how simvastatin exerts its neuroprotective role. We hypothesize that this reduction may be due to protein clearance.
Collapse
|
10
|
Janas T, Sapoń K, Stowell MHB, Janas T. Selection of Membrane RNA Aptamers to Amyloid Beta Peptide: Implications for Exosome-Based Antioxidant Strategies. Int J Mol Sci 2019; 20:ijms20020299. [PMID: 30642129 PMCID: PMC6359565 DOI: 10.3390/ijms20020299] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 12/28/2018] [Accepted: 01/07/2019] [Indexed: 12/12/2022] Open
Abstract
The distribution of amyloid beta peptide 42 (Aβ42) between model exosomal membranes and a buffer solution was measured. The model membranes contained liquid-ordered regions or phosphatidylserine. Results demonstrated that up to ca. 20% of amyloid peptide, generated in the plasma (or intracellular) membrane as a result of proteolytic cleavage of amyloid precursor proteins by β- and γ-secretases, can stay within the membrane milieu. The selection of RNA aptamers that bind to Aβ42 incorporated into phosphatidylserine-containing liposomal membranes was performed using the selection-amplification (SELEX) method. After eight selection cycles, the pool of RNA aptamers was isolated and its binding to Aβ42-containing membranes was demonstrated using the gel filtration method. Since membranes can act as a catalytic surface for Aβ42 aggregation, these RNA aptamers may inhibit the formation of toxic amyloid aggregates that can permeabilize cellular membranes or disrupt membrane receptors. Strategies are proposed for using functional exosomes, loaded with RNA aptamers specific to membrane Aβ42, to reduce the oxidative stress in Alzheimer's disease and Down's syndrome.
Collapse
Affiliation(s)
- Teresa Janas
- Institute of Biotechnology, University of Opole, Kominka 6, 45-032 Opole, Poland.
| | - Karolina Sapoń
- Institute of Biotechnology, University of Opole, Kominka 6, 45-032 Opole, Poland.
| | - Michael H B Stowell
- Department of MCD Biology, University of Colorado, Boulder, CO 80309, USA.
- Mechanical Engineering, University of Colorado, Boulder, CO 80309, USA.
| | - Tadeusz Janas
- Institute of Biotechnology, University of Opole, Kominka 6, 45-032 Opole, Poland.
- Department of MCD Biology, University of Colorado, Boulder, CO 80309, USA.
| |
Collapse
|
11
|
Houang J, Perrone GG, Pedrinazzi C, Longo L, Mawad D, Boughton PC, Ruys AJ, Lauto A. Genetic Tolerance to Rose Bengal Photodynamic Therapy and Antifungal Clinical Application for Onychomycosis. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800105] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Jessica Houang
- School of Aerospace; Mechanical and Mechatronic Engineering; University of Sydney; Sydney NSW 2006 Australia
| | - Gabriel G. Perrone
- School of Science and Health; Western Sydney University; Penrith NSW 2751 Australia
| | | | - Leonardo Longo
- School of Medicine; University of Siena; 53100 Siena Italy
| | - Damia Mawad
- School of Materials Science and Engineering; University of New South Wales; Sydney NSW 2052 Australia
- Australian Centre for NanoMedicine and ARC Centre of Excellence in Convergent BioNano Science and Technology; University of New South Wales; Sydney NSW 2052 Australia
- Centre for Advanced Macromolecular Design; University of New South Wales; Sydney NSW 2052 Australia
| | - Philip C. Boughton
- School of Aerospace; Mechanical and Mechatronic Engineering; University of Sydney; Sydney NSW 2006 Australia
| | - Andrew J. Ruys
- School of Aerospace; Mechanical and Mechatronic Engineering; University of Sydney; Sydney NSW 2006 Australia
| | - Antonio Lauto
- School of Science and Health; Western Sydney University; Penrith NSW 2751 Australia
- Biomedical Engineering and Neuroscience Research Group; The MARCS Institute; Western Sydney University; Penrith NSW 2751 Australia
- School of Medicine; Western Sydney University; Penrith NSW 2750 Australia
| |
Collapse
|
12
|
Chuartzman SG, Schuldiner M. Database for High Throughput Screening Hits (dHITS): a simple tool to retrieve gene specific phenotypes from systematic screens done in yeast. Yeast 2018; 35:477-483. [PMID: 29574976 PMCID: PMC6055851 DOI: 10.1002/yea.3312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 03/04/2018] [Accepted: 03/07/2018] [Indexed: 12/21/2022] Open
Abstract
In the last decade several collections of Saccharomyces cerevisiae yeast strains have been created. In these collections every gene is modified in a similar manner such as by a deletion or the addition of a protein tag. Such libraries have enabled a diversity of systematic screens, giving rise to large amounts of information regarding gene functions. However, often papers describing such screens focus on a single gene or a small set of genes and all other loci affecting the phenotype of choice (‘hits’) are only mentioned in tables that are provided as supplementary material and are often hard to retrieve or search. To help unify and make such data accessible, we have created a Database of High Throughput Screening Hits (dHITS). The dHITS database enables information to be obtained about screens in which genes of interest were found as well as the other genes that came up in that screen – all in a readily accessible and downloadable format. The ability to query large lists of genes at the same time provides a platform to easily analyse hits obtained from transcriptional analyses or other screens. We hope that this platform will serve as a tool to facilitate investigation of protein functions to the yeast community.
Collapse
Affiliation(s)
- Silvia G Chuartzman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, 7610001, Israel
| |
Collapse
|
13
|
Tardiff DF, Brown LE, Yan X, Trilles R, Jui NT, Barrasa MI, Caldwell KA, Caldwell GA, Schaus SE, Lindquist S. Dihydropyrimidine-Thiones and Clioquinol Synergize To Target β-Amyloid Cellular Pathologies through a Metal-Dependent Mechanism. ACS Chem Neurosci 2017; 8:2039-2055. [PMID: 28628299 PMCID: PMC5705239 DOI: 10.1021/acschemneuro.7b00187] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The lack of therapies for neurodegenerative diseases arises from our incomplete understanding of their underlying cellular toxicities and the limited number of predictive model systems. It is critical that we develop approaches to identify novel targets and lead compounds. Here, a phenotypic screen of yeast proteinopathy models identified dihydropyrimidine-thiones (DHPM-thiones) that selectively rescued the toxicity caused by β-amyloid (Aβ), the peptide implicated in Alzheimer's disease. Rescue of Aβ toxicity by DHPM-thiones occurred through a metal-dependent mechanism of action. The bioactivity was distinct, however, from that of the 8-hydroxyquinoline clioquinol (CQ). These structurally dissimilar compounds strongly synergized at concentrations otherwise not competent to reduce toxicity. Cotreatment ameliorated Aβ toxicity by reducing Aβ levels and restoring functional vesicle trafficking. Notably, these low doses significantly reduced deleterious off-target effects caused by CQ on mitochondria at higher concentrations. Both single and combinatorial treatments also reduced death of neurons expressing Aβ in a nematode, indicating that DHPM-thiones target a conserved protective mechanism. Furthermore, this conserved activity suggests that expression of the Aβ peptide causes similar cellular pathologies from yeast to neurons. Our identification of a new cytoprotective scaffold that requires metal-binding underscores the critical role of metal phenomenology in mediating Aβ toxicity. Additionally, our findings demonstrate the valuable potential of synergistic compounds to enhance on-target activities, while mitigating deleterious off-target effects. The identification and prosecution of synergistic compounds could prove useful for developing AD therapeutics where combination therapies may be required to antagonize diverse pathologies.
Collapse
Affiliation(s)
- Daniel F. Tardiff
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, United States
| | - Lauren E. Brown
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts 02215, United States
| | - Xiaohui Yan
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Richard Trilles
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts 02215, United States
| | - Nathan T. Jui
- Department of Chemistry, MIT, Cambridge, Massachusetts 02139, United States
| | - M. Inmaculada Barrasa
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, United States
| | - Kim A. Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Guy A. Caldwell
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, Alabama 35487, United States
| | - Scott E. Schaus
- Department of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts 02215, United States
| | - Susan Lindquist
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, United States
- Department of Biology, MIT, Cambridge, Massachusetts 02139, United States
- Howard Hughes Medical Institute, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
14
|
Chen X, Bisschops MMM, Agarwal NR, Ji B, Shanmugavel KP, Petranovic D. Interplay of Energetics and ER Stress Exacerbates Alzheimer's Amyloid-β (Aβ) Toxicity in Yeast. Front Mol Neurosci 2017; 10:232. [PMID: 28798664 PMCID: PMC5529408 DOI: 10.3389/fnmol.2017.00232] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/06/2017] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegeneration. Oligomers of amyloid-β peptides (Aβ) are thought to play a pivotal role in AD pathogenesis, yet the mechanisms involved remain unclear. Two major isoforms of Aβ associated with AD are Aβ40 and Aβ42, the latter being more toxic and prone to form oligomers. Here, we took a systems biology approach to study two humanized yeast AD models which expressed either Aβ40 or Aβ42 in bioreactor cultures. Strict control of oxygen availability and culture pH, strongly affected chronological lifespan and reduced variations during cell growth. Reduced growth rates and biomass yields were observed upon Aβ42 expression, indicating a redirection of energy from growth to maintenance. Quantitative physiology analyses furthermore revealed reduced mitochondrial functionality and ATP generation in Aβ42 expressing cells, which matched with observed aberrant mitochondrial structures. Genome-wide expression level analysis showed that Aβ42 expression triggered strong ER stress and unfolded protein responses. Equivalent expression of Aβ40, however, induced only mild ER stress, which resulted in hardly affected physiology. Using AD yeast models in well-controlled cultures strengthened our understanding on how cells translate different Aβ toxicity signals into particular cell fate programs, and further enhance their potential as a discovery platform to identify possible therapies.
Collapse
Affiliation(s)
- Xin Chen
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of TechnologyGothenburg, Sweden
| | - Markus M. M. Bisschops
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of TechnologyGothenburg, Sweden
| | - Nisha R. Agarwal
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of TechnologyGothenburg, Sweden
| | - Boyang Ji
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of TechnologyGothenburg, Sweden
| | - Kumaravel P. Shanmugavel
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of TechnologyGothenburg, Sweden
| | - Dina Petranovic
- Division of Systems and Synthetic Biology, Department of Biology and Biological Engineering, Chalmers University of TechnologyGothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of TechnologyGothenburg, Sweden
| |
Collapse
|
15
|
Exploring the power of yeast to model aging and age-related neurodegenerative disorders. Biogerontology 2016; 18:3-34. [PMID: 27804052 DOI: 10.1007/s10522-016-9666-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/24/2016] [Indexed: 12/12/2022]
Abstract
Aging is a multifactorial process determined by molecular, cellular and systemic factors and it is well established that advancing age is a leading risk factor for several neurodegenerative diseases. In fact, the close association of aging and neurodegenerative disorders has placed aging as the greatest social and economic challenge of the 21st century, and age-related diseases have also become a key priority for countries worldwide. The growing need to better understand both aging and neurodegenerative processes has led to the development of simple eukaryotic models amenable for mechanistic studies. Saccharomyces cerevisiae has proven to be an unprecedented experimental model to study the fundamental aspects of aging and to decipher the intricacies of neurodegenerative disorders greatly because the molecular mechanisms underlying these processes are evolutionarily conserved from yeast to human. Moreover, yeast offers several methodological advantages allowing a rapid and relatively easy way of establishing gene-protein-function associations. Here we review different aging theories, common cellular pathways driving aging and neurodegenerative diseases and discuss the major contributions of yeast to the state-of-art knowledge in both research fields.
Collapse
|
16
|
Neuroprotective Effects of the Herbal Formula B401 in Both Cell and Mouse Models of Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:1939052. [PMID: 27761145 PMCID: PMC5059538 DOI: 10.1155/2016/1939052] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 07/26/2016] [Indexed: 11/27/2022]
Abstract
In this study, we have reported the herbal formula B401 that has neuroprotective effects via multifunction, multitarget characteristics. It is possible that the herbal formula B401 may also provide new insights for AD. Here, we studied protective effects in the Tet-On Aβ42-GFP SH-SY5Y cell model and the APP/PS1/Tau triple transgenic mouse model by the herbal formula B401. In in vitro experiments, we showed that the herbal formula B401 treatment effectively reduces glutamate-induced excitotoxicity and acetylcholinesterase activity in Tet-On Aβ42-GFP SH-SY5Y cells. In in vivo experiments, we found that oral B401 treatment effectively ameliorates neurocognitive dysfunctions of 3× Tg-AD mice via motor and cognitive behavior tests. By using magnetic resonance imaging, moorFLPI instruments, and chemiluminescence methods, we reported that oral B401 treatment effectively alleviates brain atrophy, improves subcutaneous blood flow, and reduces blood ROS in 3× Tg-AD mice. As observed from results of immunohistochemistry staining and western blotting, we found that oral B401 treatment significantly enhances expressions of neuroprotective proteins, while reducing expressions of AD derived proteins such as amyloid beta, phosphorylated Tau, neurofibrillary tangles, and 3-nitrotyrosine in the brain of 3× Tg-AD mice. Thus, the herbal formula B401 may have the potential to be developed into optimum TCM for AD patients.
Collapse
|
17
|
Chen X, Petranovic D. Role of frameshift ubiquitin B protein in Alzheimer's disease. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2016; 8:300-13. [DOI: 10.1002/wsbm.1340] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/10/2016] [Accepted: 03/19/2016] [Indexed: 12/20/2022]
Affiliation(s)
- Xin Chen
- Systems and Synthetic Biology, Department of Biology and Biological Engineering; Chalmers University of Technology; Göteborg Sweden
| | - Dina Petranovic
- Systems and Synthetic Biology, Department of Biology and Biological Engineering; Chalmers University of Technology; Göteborg Sweden
- Novo Nordisk Foundation Center for Biosustainability; Chalmers University of Technology; Göteborg Sweden
| |
Collapse
|
18
|
Ochiishi T, Doi M, Yamasaki K, Hirose K, Kitamura A, Urabe T, Hattori N, Kinjo M, Ebihara T, Shimura H. Development of new fusion proteins for visualizing amyloid-β oligomers in vivo. Sci Rep 2016; 6:22712. [PMID: 26982553 PMCID: PMC4793674 DOI: 10.1038/srep22712] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/18/2016] [Indexed: 12/30/2022] Open
Abstract
The intracellular accumulation of amyloid-β (Aβ) oligomers critically contributes to disease progression in Alzheimer’s disease (AD) and can be the potential target of AD therapy. Direct observation of molecular dynamics of Aβ oligomers in vivo is key for drug discovery research, however, it has been challenging because Aβ aggregation inhibits the fluorescence from fusion proteins. Here, we developed Aβ1-42-GFP fusion proteins that are oligomerized and visualize their dynamics inside cells even when aggregated. We examined the aggregation states of Aβ-GFP fusion proteins using several methods and confirmed that they did not assemble into fibrils, but instead formed oligomers in vitro and in live cells. By arranging the length of the liker between Aβ and GFP, we generated two fusion proteins with “a long-linker” and “a short-linker”, and revealed that the aggregation property of fusion proteins can be evaluated by measuring fluorescence intensities using rat primary culture neurons transfected with Aβ-GFP plasmids and Aβ-GFP transgenic C. elegans. We found that Aβ-GFP fusion proteins induced cell death in COS7 cells. These results suggested that novel Aβ-GFP fusion proteins could be utilized for studying the physiological functions of Aβ oligomers in living cells and animals, and for drug screening by analyzing Aβ toxicity.
Collapse
Affiliation(s)
- Tomoyo Ochiishi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Motomichi Doi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Kazuhiko Yamasaki
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Keiko Hirose
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Akira Kitamura
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
| | - Takao Urabe
- Department of Neurology, Juntendo University Urayasu Hospital, 2-1-1, Tomioka, Urayasu, Chiba 279-0021, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Masataka Kinjo
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
| | - Tatsuhiko Ebihara
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Hideki Shimura
- Department of Neurology, Juntendo University Urayasu Hospital, 2-1-1, Tomioka, Urayasu, Chiba 279-0021, Japan
| |
Collapse
|
19
|
Ochiishi T, Doi M, Yamasaki K, Hirose K, Kitamura A, Urabe T, Hattori N, Kinjo M, Ebihara T, Shimura H. Development of new fusion proteins for visualizing amyloid-β oligomers in vivo. Sci Rep 2016. [PMID: 26982553 DOI: 10.1038/srep22712(2016)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The intracellular accumulation of amyloid-β (Aβ) oligomers critically contributes to disease progression in Alzheimer's disease (AD) and can be the potential target of AD therapy. Direct observation of molecular dynamics of Aβ oligomers in vivo is key for drug discovery research, however, it has been challenging because Aβ aggregation inhibits the fluorescence from fusion proteins. Here, we developed Aβ1-42-GFP fusion proteins that are oligomerized and visualize their dynamics inside cells even when aggregated. We examined the aggregation states of Aβ-GFP fusion proteins using several methods and confirmed that they did not assemble into fibrils, but instead formed oligomers in vitro and in live cells. By arranging the length of the liker between Aβ and GFP, we generated two fusion proteins with "a long-linker" and "a short-linker", and revealed that the aggregation property of fusion proteins can be evaluated by measuring fluorescence intensities using rat primary culture neurons transfected with Aβ-GFP plasmids and Aβ-GFP transgenic C. elegans. We found that Aβ-GFP fusion proteins induced cell death in COS7 cells. These results suggested that novel Aβ-GFP fusion proteins could be utilized for studying the physiological functions of Aβ oligomers in living cells and animals, and for drug screening by analyzing Aβ toxicity.
Collapse
Affiliation(s)
- Tomoyo Ochiishi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Motomichi Doi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Kazuhiko Yamasaki
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Keiko Hirose
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Akira Kitamura
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
| | - Takao Urabe
- Department of Neurology, Juntendo University Urayasu Hospital, 2-1-1, Tomioka, Urayasu, Chiba 279-0021, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Masataka Kinjo
- Laboratory of Molecular Cell Dynamics, Faculty of Advanced Life Science, Hokkaido University, N21W11, Kita-ku, Sapporo 001-0021, Japan
| | - Tatsuhiko Ebihara
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Hideki Shimura
- Department of Neurology, Juntendo University Urayasu Hospital, 2-1-1, Tomioka, Urayasu, Chiba 279-0021, Japan
| |
Collapse
|
20
|
Wu Y, Du J, Xu G, Jiang L. The transcription factor Ace2 and its paralog Swi5 regulate ethanol production during static fermentation through their targets Cts1 and Rps4a inSaccharomyces cerevisiae. FEMS Yeast Res 2016; 16:fow022. [DOI: 10.1093/femsyr/fow022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2016] [Indexed: 12/26/2022] Open
|
21
|
Torres NP, Ho B, Brown GW. High-throughput fluorescence microscopic analysis of protein abundance and localization in budding yeast. Crit Rev Biochem Mol Biol 2016; 51:110-9. [PMID: 26893079 DOI: 10.3109/10409238.2016.1145185] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Proteins directly carry out and regulate cellular functions. As a result, changes in protein levels within a cell directly influence cellular processes. Similarly, it is intuitive that the intracellular localization of proteins is a key component of their functionality. Optimal activity is achieved by a combination of protein concentration, co-compartmentalization with substrates, co-factors and regulators and sequestration from deleterious locales. The proteome within a cell is highly dynamic and changes in response to different environmental conditions. High-throughput microscopic analysis in the budding yeast Saccharomyces cerevisiae has afforded proteome-wide views of protein organization in living cells, and of how protein abundance and location is regulated and remodeled in response to stress.
Collapse
Affiliation(s)
- Nikko P Torres
- a Department of Biochemistry and Donnelly Centre , University of Toronto , Toronto , Ontario , Canada
| | - Brandon Ho
- a Department of Biochemistry and Donnelly Centre , University of Toronto , Toronto , Ontario , Canada
| | - Grant W Brown
- a Department of Biochemistry and Donnelly Centre , University of Toronto , Toronto , Ontario , Canada
| |
Collapse
|